SLAS DiscoveryPub Date : 2023-10-01DOI: 10.1016/j.slasd.2023.08.009
Gregory P. Way , Heba Sailem , Steven Shave , Richard Kasprowicz , Neil O. Carragher
{"title":"Evolution and impact of high content imaging","authors":"Gregory P. Way , Heba Sailem , Steven Shave , Richard Kasprowicz , Neil O. Carragher","doi":"10.1016/j.slasd.2023.08.009","DOIUrl":"10.1016/j.slasd.2023.08.009","url":null,"abstract":"<div><p>The field of high content imaging has steadily evolved and expanded substantially across many industry and academic research institutions since it was first described in the early 1990′s. High content imaging refers to the automated acquisition and analysis of microscopic images from a variety of biological sample types. Integration of high content imaging microscopes with multiwell plate handling robotics enables high content imaging to be performed at scale and support medium- to high-throughput screening of pharmacological, genetic and diverse environmental perturbations upon complex biological systems ranging from 2D cell cultures to 3D tissue organoids to small model organisms. In this perspective article the authors provide a collective view on the following key discussion points relevant to the evolution of high content imaging:</p><p>• Evolution and impact of high content imaging: An academic perspective</p><p>• Evolution and impact of high content imaging: An industry perspective</p><p>• Evolution of high content image analysis</p><p>• Evolution of high content data analysis pipelines towards multiparametric and phenotypic profiling applications</p><p>• The role of data integration and multiomics</p><p>• The role and evolution of image data repositories and sharing standards</p><p>• Future perspective of high content imaging hardware and software</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10541226","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-10-01DOI: 10.1016/j.slasd.2023.07.003
Sean M. McCarty , Martin C. Clasby , Jonathan Z. Sexton
{"title":"Automated high-throughput, high-content 3D imaging of intact pancreatic islets","authors":"Sean M. McCarty , Martin C. Clasby , Jonathan Z. Sexton","doi":"10.1016/j.slasd.2023.07.003","DOIUrl":"10.1016/j.slasd.2023.07.003","url":null,"abstract":"<div><p>Diabetes poses a global health crisis affecting individuals across age groups and backgrounds, with a prevalence estimate of 700 million people worldwide by 2045. Current therapeutic strategies primarily rely on insulin therapy or hypoglycemic agents, which fail to address the root cause of the disease - the loss of pancreatic insulin-producing beta-cells. Therefore, bioassays that recapitulate intact islets are needed to enable drug discovery for beta-cell replenishment, protection from beta-cell loss, and islet-cell interactions. Standard cancer insulinoma beta-cell lines MIN6 and INS-1 have been used to interrogate beta-cell metabolic pathways and function but are not suitable for studying proliferative effects. Screening using primary human/rodent intact islets offers a higher level of physiological relevance to enhance diabetes drug discovery and development. However, the 3-dimensionality of intact islets have presented challenges in developing robust, high-throughput assays to detect beta-cell proliferative effects. Established methods rely on either dissociated islet cells plated in 2D monolayer cultures for imaging or reconstituted pseudo-islets formed in round bottom plates to achieve homogeneity. These approaches have significant limitations due to the islet cell dispersion process. To address these limitations, we have developed a robust, intact <em>ex vivo</em> pancreatic islet bioassay in 384-well format that is capable of detecting diabetes-relevant endpoints including beta-cell proliferation, chemoprotection, and islet spatial morphometrics.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9941430","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-10-01DOI: 10.1016/j.slasd.2023.08.001
Ashley T. Fancher , Yun Hua , David A. Close , Wei Xu , Lee A. McDermott , Christopher J. Strock , Ulises Santiago , Carlos J. Camacho , Paul A. Johnston
{"title":"Characterization of allosteric modulators that disrupt androgen receptor co-activator protein-protein interactions to alter transactivation–Drug leads for metastatic castration resistant prostate cancer","authors":"Ashley T. Fancher , Yun Hua , David A. Close , Wei Xu , Lee A. McDermott , Christopher J. Strock , Ulises Santiago , Carlos J. Camacho , Paul A. Johnston","doi":"10.1016/j.slasd.2023.08.001","DOIUrl":"10.1016/j.slasd.2023.08.001","url":null,"abstract":"<div><p>Three series of compounds were prioritized from a high content screening campaign that identified molecules that blocked dihydrotestosterone (DHT) induced formation of Androgen Receptor (AR) protein-protein interactions (PPIs) with the Transcriptional Intermediary Factor 2 (TIF2) coactivator and also disrupted preformed AR-TIF2 PPI complexes; the hydrobenzo-oxazepins (S1), thiadiazol-5-piperidine-carboxamides (S2), and phenyl-methyl-indoles (S3). Compounds from these series inhibited AR PPIs with TIF2 and SRC-1, another p160 coactivator, in mammalian 2-hybrid assays and blocked transcriptional activation in reporter assays driven by full length AR or AR-V7 splice variants. Compounds inhibited the growth of five prostate cancer cell lines, with many exhibiting differential cytotoxicity towards AR positive cell lines. Representative compounds from the 3 series substantially reduced both endogenous and DHT-enhanced expression and secretion of the prostate specific antigen (PSA) cancer biomarker in the C4–2 castration resistant prostate cancer (CRPC) cell line. The comparatively weak activities of series compounds in the H<sup>3</sup>-DHT and/or TIF2 box 3 LXXLL-peptide binding assays to the recombinant ligand binding domain of AR suggest that direct antagonism at the orthosteric ligand binding site or AF-2 surface respectively are unlikely mechanisms of action. Cellular enhanced thermal stability assays (CETSA) indicated that compounds engaged AR and reduced the maximum efficacy and right shifted the EC<sub>50</sub> of DHT-enhanced AR thermal stabilization consistent with the effects of negative allosteric modulators. Molecular docking of potent representative hits from each series to AR structures suggest that S1–1 and S2–6 engage a novel binding pocket (BP-1) adjacent to the orthosteric ligand binding site, while S3–11 occupies the AR binding function 3 (BF-3) allosteric pocket. Hit binding poses indicate spaces and residues adjacent to the BP-1 and BF-3 pockets that will be exploited in future medicinal chemistry optimization studies. Small molecule allosteric modulators that prevent/disrupt AR PPIs with coactivators like TIF2 to alter transcriptional activation in the presence of orthosteric agonists might evade the resistance mechanisms to existing prostate cancer drugs and provide novel starting points for medicinal chemistry lead optimization and future development into therapies for metastatic CRPC.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10011608","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-10-01DOI: 10.1016/j.slasd.2023.08.003
Yun Huang , Aneta Drakul , Jasmeet Sidhu , Kerstin K. Rauwolf , James Kim , Beat Bornhauser , Jean-Pierre Bourquin
{"title":"MSC.sensor: Capturing cancer cell interactions with stroma for functional profiling","authors":"Yun Huang , Aneta Drakul , Jasmeet Sidhu , Kerstin K. Rauwolf , James Kim , Beat Bornhauser , Jean-Pierre Bourquin","doi":"10.1016/j.slasd.2023.08.003","DOIUrl":"10.1016/j.slasd.2023.08.003","url":null,"abstract":"<div><p>Mesenchymal stromal cells (MSCs) contribute to the microenvironment regulating normal and malignant hematopoiesis, and thus may support subpopulations of cancer cells to escape therapeutic pressure. Here, we engineered bone marrow MSCs to express a synthetic CD19-sensor receptor to detect and display interacting primary CD19+ leukemia cells in coculture. This implementation provides a versatile platform facilitating <em>ex vivo</em> drug response profiling of primary CD19+ leukemia cells in coculture with high-sensitivity and scalability.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10025756","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Novel beta-glucocerebrosidase chaperone compounds identified from cell-based screening reduce pathologically accumulated glucosylsphingosine in iPS-derived neuronal cells","authors":"Yusuke Naito, Sou Sakamoto, Takuto Kojima, Misaki Homma, Maiko Tanaka, Hideki Matsui","doi":"10.1016/j.slasd.2023.06.002","DOIUrl":"10.1016/j.slasd.2023.06.002","url":null,"abstract":"<div><p>The beta-glucocerebrosidase (<em>GBA1</em>) gene encodes the lysosomal beta-glucocerebrosidase (GCase) that metabolizes the lipids glucosylceramide (GlcCer) and glucosylsphingosine (GlcSph). Biallelic loss-of-function mutations in <em>GBA1</em> such as L444P cause Gaucher disease (GD), which is the most prevalent lysosomal storage disease and is histopathologically characterized by abnormal accumulation of the GCase substrates GlcCer and GlcSph. GD with neurological symptoms is associated with severe mutations in the <em>GBA1</em> gene, most of which cause impairment in the process of GCase trafficking to lysosomes. Given that recombinant GCase protein cannot cross the blood-brain barrier due to its high molecular weight, it is invaluable to develop a brain-penetrant small-molecule pharmacological chaperone as a viable therapeutic strategy to boost GCase activity in the central nervous system.</p><p>Despite considerable efforts to screen potent GCase activators/chaperones, cell-free assays using recombinant GCase protein have yielded compounds with only marginal efficacy and micromolar EC<sub>50</sub> that would not have sufficient clinical efficacy or an acceptable safety margin. Therefore, we utilized a fluorescence-labeled GCase suicide inhibitor, MDW933, to directly monitor lysosomal GCase activity and performed a cell-based screening in fibroblasts from a GD patient with homozygotic L444P mutations. Here, we identified novel compounds that increase the fluorescence signal from labeled GCase with L444P mutations in a dose-dependent manner. Secondary assays using an artificial cell-permeable lysosomal GCase substrate also demonstrated that the identified compounds augment lysosomal GCase L444P in the fibroblast. Moreover, those compounds increased the total GCase L444P protein levels, suggesting the pharmacological chaperone-like mechanism of action. To further elucidate the effect of the compounds on the endogenous GCase substrate GlcSph, we generated iPSC-derived dopaminergic neurons with a <em>GBA1</em> L444P mutation that exhibit GlcSph accumulation in vitro. Importantly, the identified compounds reduce GlcSph in iPSC-derived dopaminergic neurons with a <em>GBA1</em> L444P mutation, indicating that the increase in lysosomal GCase resulting from application of the compounds leads to the clearance of pathologically-accumulated GlcSph. Together, our findings pave the way for developing potent and efficacious GCase chaperone compounds as a potential therapeutic approach for neurological GD.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9814762","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-10-01DOI: 10.1016/j.slasd.2023.08.004
Jeff Winchell, Gabriel Comolet, Geoff Buckley-Herd, Dillion Hutson, Neeloy Bose, Daniel Paull, Bianca Migliori
{"title":"FocA: A deep learning tool for reliable, near-real-time imaging focus analysis in automated cell assay pipelines","authors":"Jeff Winchell, Gabriel Comolet, Geoff Buckley-Herd, Dillion Hutson, Neeloy Bose, Daniel Paull, Bianca Migliori","doi":"10.1016/j.slasd.2023.08.004","DOIUrl":"10.1016/j.slasd.2023.08.004","url":null,"abstract":"<div><p>The increasing use of automation in cellular assays and cell culture presents significant opportunities to enhance the scale and throughput of imaging assays, but to do so, reliable data quality and consistency are critical. Realizing the full potential of automation will thus require the design of robust analysis pipelines that span the entire workflow in question. Here we present FocA, a deep learning tool that, in near real-time, identifies in-focus and out-of-focus images generated on a fully automated cell biology research platform, the NYSCF Global Stem Cell Array®. The tool is trained on small patches of downsampled images to maximize computational efficiency without compromising accuracy, and optimized to make sure no sub-quality images are stored and used in downstream analyses. The tool automatically generates balanced and maximally diverse training sets to avoid bias. The resulting model correctly identifies 100% of out-of-focus and 98% of in-focus images in under 4 s per 96-well plate, and achieves this result even in heavily downsampled data (∼30 times smaller than native resolution). Integrating the tool into automated workflows minimizes the need for human verification as well as the collection and usage of low-quality data. FocA thus offers a solution to ensure reliable image data hygiene and improve the efficiency of automated imaging workflows using minimal computational resources.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10066166","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-10-01DOI: 10.1016/j.slasd.2023.10.001
Neil O. Carragher , Judi Wardwell-Swanson , Gregory P. Way
{"title":"High-content imaging 2023: A joint special collection with the society for biomolecular imaging","authors":"Neil O. Carragher , Judi Wardwell-Swanson , Gregory P. Way","doi":"10.1016/j.slasd.2023.10.001","DOIUrl":"10.1016/j.slasd.2023.10.001","url":null,"abstract":"","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41223020","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-09-01DOI: 10.1016/j.slasd.2023.05.003
Elizabeth R. Sharlow
{"title":"Emerging drug discovery ecosystems","authors":"Elizabeth R. Sharlow","doi":"10.1016/j.slasd.2023.05.003","DOIUrl":"10.1016/j.slasd.2023.05.003","url":null,"abstract":"","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10300625","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-09-01DOI: 10.1016/j.slasd.2023.03.001
Ali Andalibi , Remi Veneziano , Mikell Paige , Michael Buschmann , Amanda Haymond , Virginia Espina , Alessandra Luchini , Lance Liotta , Barney Bishop , Monique Van Hoek
{"title":"Drug discovery efforts at George Mason University","authors":"Ali Andalibi , Remi Veneziano , Mikell Paige , Michael Buschmann , Amanda Haymond , Virginia Espina , Alessandra Luchini , Lance Liotta , Barney Bishop , Monique Van Hoek","doi":"10.1016/j.slasd.2023.03.001","DOIUrl":"10.1016/j.slasd.2023.03.001","url":null,"abstract":"<div><p>With over 39,000 students, and research expenditures in excess of $200 million, George Mason University (GMU) is the largest R1 (Carnegie Classification of very high research activity) university in Virginia. Mason scientists have been involved in the discovery and development of novel diagnostics and therapeutics in areas as diverse as infectious diseases and cancer. Below are highlights of the efforts being led by Mason researchers in the drug discovery arena.</p><p>To enable targeted cellular delivery, and non-biomedical applications, Veneziano and colleagues have developed a synthesis strategy that enables the design of self-assembling DNA nanoparticles (DNA origami) with prescribed shape and size in the 10 to 100 nm range. The nanoparticles can be loaded with molecules of interest such as drugs, proteins and peptides, and are a promising new addition to the drug delivery platforms currently in use. The investigators also recently used the DNA origami nanoparticles to fine tune the spatial presentation of immunogens to study the impact on B cell activation. These studies are an important step towards the rational design of vaccines for a variety of infectious agents.</p><p>To elucidate the parameters for optimizing the delivery efficiency of lipid nanoparticles (LNPs), Buschmann, Paige and colleagues have devised methods for predicting and experimentally validating the pKa of LNPs based on the structure of the ionizable lipids used to formulate the LNPs. These studies may pave the way for the development of new LNP delivery vehicles that have reduced systemic distribution and improved endosomal release of their cargo post administration.</p><p>To better understand protein-protein interactions and identify potential drug targets that disrupt such interactions, Luchini and colleagues have developed a methodology that identifies contact points between proteins using small molecule dyes. The dye molecules noncovalently bind to the accessible surfaces of a protein complex with very high affinity, but are excluded from contact regions. When the complex is denatured and digested with trypsin, the exposed regions covered by the dye do not get cleaved by the enzyme, whereas the contact points are digested. The resulting fragments can then be identified using mass spectrometry. The data generated can serve as the basis for designing small molecules and peptides that can disrupt the formation of protein complexes involved in disease processes. For example, using peptides based on the interleukin 1 receptor accessory protein (IL-1RAcP), Luchini, Liotta, Paige and colleagues disrupted the formation of IL-1/IL-R/IL-1RAcP complex and demonstrated that the inhibition of complex formation reduced the inflammatory response to IL-1B.</p><p>Working on the discovery of novel antimicrobial agents, Bishop, van Hoek and colleagues have discovered a number of antimicrobial peptides from reptiles and other species. DRGN-1, is a synthetic peptide based on a his","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10660919","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2023-09-01DOI: 10.1016/j.slasd.2022.10.002
Elizabeth R. Sharlow , Danielle C. Llaneza , William E. Grever , Garnett A. Mingledorff , Anna J. Mendelson , George S. Bloom , John S. Lazo
{"title":"High content screening miniaturization and single cell imaging of mature human feeder layer-free iPSC-derived neurons","authors":"Elizabeth R. Sharlow , Danielle C. Llaneza , William E. Grever , Garnett A. Mingledorff , Anna J. Mendelson , George S. Bloom , John S. Lazo","doi":"10.1016/j.slasd.2022.10.002","DOIUrl":"10.1016/j.slasd.2022.10.002","url":null,"abstract":"<div><p>Human induced pluripotent stem cell (iPSC)-derived neurons are being increasingly used for high content imaging and screening. However, iPSC-derived neuronal differentiation and maturation is time-intensive, often requiring >8 weeks. Unfortunately, the differentiating and maturing iPSC-derived neuronal cultures also tend to migrate and coalesce into ganglion-like clusters making single-cell analysis challenging, especially in miniaturized formats. Using our defined extracellular matrix and low oxygen culturing conditions for the differentiation and maturation of human cortical neurons, we further modified neuronal progenitor cell seeding densities and feeder layer-free culturing conditions in miniaturized formats (<em>i.e.</em>, 96 well) to decrease neuronal clustering, enhance single-cell identification and reduce edge effects usually observed after extended neuronal cell culture. Subsequent algorithm development refined capabilities to distinguish and identify single mature neurons, as identified by NeuN expression, from large cellular aggregates, which were excluded from image analysis. Incorporation of astrocyte conditioned medium during differentiation and maturation periods significantly increased the percentage (<em>i.e.</em>, ∼10% to ∼30%) of mature neurons (<em>i.e.</em>, NeuN+) detected at 4-weeks post-differentiation. Pilot, proof of concept studies using this optimized assay system yielded negligible edge effects and robust Z-factors in population-based as well as image-based neurotoxicity assay formats. Moreover, moxidectin, an FDA-approved drug with documented neurotoxic adverse effects, was identified as a hit using both screening formats. This miniaturized, feeder layer-free format and image analysis algorithm provides a foundational imaging and screening platform, which enables quantitative single-cell analysis of differentiated human neurons.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2023-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/e0/5a/nihms-1878288.PMC10119332.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10294727","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}